Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 13: 1054477, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36466930

RESUMEN

Cryptococcal meningitis is the most common cause of meningitis among HIV/AIDS patients in sub-Saharan Africa, and worldwide causes over 223,000 cases leading to more than 181,000 annual deaths. Usually, the fungus gets inhaled into the lungs where the initial interactions occur with pulmonary phagocytes such as dendritic cells and macrophages. Following phagocytosis, the pathogen can be killed or can replicate intracellularly. Previous studies in mice showed that different subsets of these innate immune cells can either be antifungal or permissive for intracellular fungal growth. Our studies tested phagocytic antigen-presenting cell (APC) subsets from the human lung against C. neoformans. Human bronchoalveolar lavage was processed for phagocytic APCs and incubated with C. neoformans for two hours to analyze the initial interactions and fate of the fungus, living or killed. Results showed all subsets (3 macrophage and 3 dendritic cell subsets) interacted with the fungus, and both living and killed morphologies were discernable within the subsets using imaging flow cytometry. Single cell RNA-seq identified several different clusters of cells which more closely related to interactions with C. neoformans and its protective capacity against the pathogen rather than discrete cellular subsets. Differential gene expression analyses identified several changes in the innate immune cell's transcriptome as it kills the fungus including increases of TNF-α (TNF) and the switch to using fatty acid metabolism by upregulation of the gene FABP4. Also, increases of TNF-α correlated to cryptococcal interactions and uptake. Together, these analyses implicated signaling networks that regulate expression of many different genes - both metabolic and immune - as certain clusters of cells mount a protective response and kill the pathogen. Future studies will examine these genes and networks to understand the exact mechanism(s) these phagocytic APC subsets use to kill C. neoformans in order to develop immunotherapeutic strategies to combat this deadly disease.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Humanos , Animales , Ratones , Presentación de Antígeno , Factor de Necrosis Tumoral alfa , Fagocitos
2.
Toxins (Basel) ; 12(7)2020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-32698436

RESUMEN

Bacillus anthracis, the causative agent of inhalation anthrax, is a serious concern as a bioterrorism weapon. The vegetative form produces two exotoxins: Lethal toxin (LT) and edema toxin (ET). We recently characterized and compared six human airway and alveolar-resident phagocyte (AARP) subsets at the transcriptional and functional levels. In this study, we examined the effects of LT and ET on these subsets and human leukocytes. AARPs and leukocytes do not express high levels of the toxin receptors, tumor endothelium marker-8 (TEM8) and capillary morphogenesis protein-2 (CMG2). Less than 20% expressed surface TEM8, while less than 15% expressed CMG2. All cell types bound or internalized protective antigen, the common component of the two toxins, in a dose-dependent manner. Most protective antigen was likely internalized via macropinocytosis. Cells were not sensitive to LT-induced apoptosis or necrosis at concentrations up to 1000 ng/mL. However, toxin exposure inhibited B. anthracis spore internalization. This inhibition was driven primarily by ET in AARPs and LT in leukocytes. These results support a model of inhalation anthrax in which spores germinate and produce toxins. ET inhibits pathogen phagocytosis by AARPs, allowing alveolar escape. In late-stage disease, LT inhibits phagocytosis by leukocytes, allowing bacterial replication in the bloodstream.


Asunto(s)
Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Leucocitos/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Pinocitosis/efectos de los fármacos , Adolescente , Adulto , Anciano , Animales , Apoptosis/efectos de los fármacos , Bacillus anthracis/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Leucocitos/metabolismo , Leucocitos/patología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Masculino , Ratones , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Necrosis , Células RAW 264.7 , Receptores de Superficie Celular/metabolismo , Receptores de Péptidos/metabolismo , Esporas Bacterianas/metabolismo , Adulto Joven
3.
Microb Pathog ; 121: 9-21, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29704667

RESUMEN

The lung is the entry site for Bacillus anthracis in inhalation anthrax, the most deadly form of the disease. Spores must escape through the alveolar epithelial cell (AEC) barrier and migrate to regional lymph nodes, germinate and enter the circulatory system to cause disease. Several mechanisms to explain alveolar escape have been postulated, and all these tacitly involve the AEC barrier. In this study, we incorporate our primary human type I AEC model, microarray and gene enrichment analysis, qRT-PCR, multiplex ELISA, and neutrophil and monocyte chemotaxis assays to study the response of AEC to B. anthracis, (Sterne) spores at 4 and 24 h post-exposure. Spore exposure altered gene expression in AEC after 4 and 24 h and differentially expressed genes (±1.3 fold, p ≤ 0.05) included CCL4/MIP-1ß (4 h), CXCL8/IL-8 (4 and 24 h) and CXCL5/ENA-78 (24 h). Gene enrichment analysis revealed that pathways involving cytokine or chemokine activity, receptor binding, and innate immune responses to infection were prominent. Microarray results were confirmed by qRT-PCR and multiplex ELISA assays. Chemotaxis assays demonstrated that spores induced the release of biologically active neutrophil and monocyte chemokines, and that CXCL8/IL-8 was the major neutrophil chemokine. The small or sub-chemotactic doses of CXCL5/ENA-78, CXCL2/GROß and CCL20/MIP-3α may contribute to chemotaxis by priming effects. These data provide the first whole transcriptomic description of the human type I AEC initial response to B. anthracis spore exposure. Taken together, our findings contribute to an increased understanding of the role of AEC in the pathogenesis of inhalational anthrax.


Asunto(s)
Células Epiteliales Alveolares/microbiología , Bacillus anthracis/patogenicidad , Quimiocinas/metabolismo , Perfilación de la Expresión Génica , Esporas Bacterianas/patogenicidad , Carbunco/genética , Carbunco/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Quimiocinas/genética , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Monocitos/metabolismo , Monocitos/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Factor Plaquetario 4/genética , Factor Plaquetario 4/metabolismo , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/metabolismo , Regulación hacia Arriba
4.
J Immunol ; 198(3): 1183-1201, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031342

RESUMEN

The respiratory system is a complex network of many cell types, including subsets of macrophages and dendritic cells that work together to maintain steady-state respiration. Owing to limitations in acquiring cells from healthy human lung, these subsets remain poorly characterized transcriptionally and phenotypically. We set out to systematically identify these subsets in human airways by developing a schema of isolating large numbers of cells by whole-lung bronchoalveolar lavage. Six subsets of phagocytic APC (HLA-DR+) were consistently observed. Aside from alveolar macrophages, subsets of Langerin+, BDCA1-CD14+, BDCA1+CD14+, BDCA1+CD14-, and BDCA1-CD14- cells were identified. These subsets varied in their ability to internalize Escherichia coli, Staphylococcus aureus, and Bacillus anthracis particles. All subsets were more efficient at internalizing S. aureus and B. anthracis compared with E. coli Alveolar macrophages and CD14+ cells were overall more efficient at particle internalization compared with the four other populations. Subsets were further separated into two groups based on their inherent capacities to upregulate surface CD83, CD86, and CCR7 expression levels. Whole-genome transcriptional profiling revealed a clade of "true dendritic cells" consisting of Langerin+, BDCA1+CD14+, and BDCA1+CD14- cells. The dendritic cell clade was distinct from a macrophage/monocyte clade, as supported by higher mRNA expression levels of several dendritic cell-associated genes, including CD1, FLT3, CX3CR1, and CCR6 Each clade, and each member of both clades, was discerned by specific upregulated genes, which can serve as markers for future studies in healthy and diseased states.


Asunto(s)
Células Dendríticas/fisiología , Pulmón/inmunología , Macrófagos Alveolares/fisiología , Macrófagos/fisiología , Adulto , Anciano , Antígenos CD/análisis , Antígenos CD1/análisis , Antígeno B7-2/análisis , Células Dendríticas/clasificación , Perfilación de la Expresión Génica , Glicoproteínas/análisis , Humanos , Inmunoglobulinas/análisis , Receptores de Lipopolisacáridos/análisis , Pulmón/microbiología , Macrófagos/clasificación , Glicoproteínas de Membrana/análisis , Persona de Mediana Edad , Antígeno CD83
5.
Microbes Infect ; 18(10): 615-626, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27320392

RESUMEN

The lung is the entry site for Bacillus anthracis in inhalation anthrax, the most deadly form of the disease. Spores escape from the alveolus to regional lymph nodes, germinate and enter the circulatory system to cause disease. The roles of carrier cells and the effects of B. anthracis toxins in this process are unclear. We used a human lung organ culture model to measure spore uptake by antigen presenting cells (APC) and alveolar epithelial cells (AEC), spore partitioning between these cells, and the effects of B. anthracis lethal toxin and protective antigen. We repeated the study in a human A549 alveolar epithelial cell model. Most spores remained unassociated with cells, but the majority of cell-associated spores were in AEC, not in APC. Spore movement was not dependent on internalization, although the location of internalized spores changed in both cell types. Spores also internalized in a non-uniform pattern. Toxins affected neither transit of the spores nor the partitioning of spores into AEC and APC. Our results support a model of spore escape from the alveolus that involves spore clustering with transient passage through intact AEC. However, subsequent transport of spores by APC from the lung to the lymph nodes may occur.


Asunto(s)
Carbunco/patología , Antígenos Bacterianos/metabolismo , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/metabolismo , Pulmón/microbiología , Ganglios Linfáticos/microbiología , Movimiento , Esporas Bacterianas/patogenicidad , Células Presentadoras de Antígenos/microbiología , Sangre/microbiología , Línea Celular , Células Epiteliales/microbiología , Humanos , Modelos Teóricos , Técnicas de Cultivo de Órganos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...